Nature - 2019.08.29

(Frankie) #1

Article reSeArcH


points (Fig. 3d and Supplementary Data). We found many previously
reported tumour-promoting factors^12 –^19 , further validating the ability


of our labelling system to faithfully capture the in vivo metastatic niche.
We also found WNT1-induced protein (WISP1)—which has been sug-


gested to act as an oncogene in breast cancer^20 —to be widely expressed
in the mCherry+ niche (Fig. 3d). Indeed, we detected upregulation of


WISP1 in both cancer and metastatic-niche cells and confirmed its
pro-metastatic activity by exogenous inhibition in vivo (Fig. 3e and


Extended Data Fig. 5a–e).
We next probed the TME for other non-immune cell types, which


might be difficult to resolve by standard techniques owing to their small
numbers. Of note, we found pathways associated with lung epithelial


cells in the metastatic-niche signature (Fig. 3f). Micro-metastases grow
embedded within the alveolar compartment of the lung, and we found


alveolar type II cells (AT2) expressing surfactant protein C (SP-C,
encoded by Sftpc) in the metastatic niche (Fig. 3g). We also found


mCherry+-niche cells expressing the epithelial cell adhesion marker
EPCAM, further supporting the presence of cells of lung parenchymal


origin (Fig. 3h, i).


Cancer-associated parenchymal cells
We found mCherry+-niche epithelial cells to have a higher prolifer-


ative activity compared to their normal lung counterparts (Fig. 4a).
Concordantly, we detected alveolar cell clusters with increased pro-


liferative activity at the metastatic borders of human breast cancer
lung metastases, suggesting that a lung parenchymal response to


metastatic growth may occur in both mouse and human (Extended


Data Fig. 6a–f). Cancer cells benefit from the presence of a lung paren-
chymal response, as freshly isolated EPCAM+ cells from naive lungs
supported the growth of actin–GFP+ MMTV–PyMT tumour cells in
our 3D-scaffold co-culture system (Fig. 4b–d). Moreover, in line with
the results shown in Fig. 2c–e, the presence of both lung neutrophils
and epithelial cells further enhanced tumour growth (Extended Data
Fig. 7a–d), highlighting the cellular complexity of the metastatic niche.
We next aimed to better define the perturbation occurring in lung
epithelial cells in the proximity of cancer cells. To contextualize their
presence among the other cellular components of the metastatic niche,
we performed single-cell RNA sequencing (scRNA-seq) of CD45− cells.
t-Distributed stochastic neighbour embedding (t-SNE) analysis of
mCherry+-niche cells identified a large stromal cluster in which dif-
ferent stromal cells could be distinguished (Fig. 4e and Extended Data
Fig. 8a–c). This is in agreement with the various known mesenchymal
cell components of the TME, as well as the characterization of differ-
ent fibroblast subsets^21 –^24. Notably, specifically in the mCherry+ niche,
Epcam-expressing epithelial cells are distributed in two clusters distin-
guished by the expression of E-cadherin (Cdh1) (Fig. 4e). We found
that only mCherry+-niche Epcam+Cdh1+ cells shared the expression
of alveolar genes^25 with unlabelled distant lung Epcam+ cells (Fig. 4f, g).
Conversely, mCherry+-niche Epcam+Cdh 1 − cells expressed both the
progenitor markers SCA1 (encoded by Ly6a) and Tm4sf1^26 –^28 (Fig. 4g).
As validation of this de-differentiated signature observed in the major-
ity of epithelial cells in the metastatic niche, reverse transcription with
quantitative PCR (RT–qPCR) of EPCAM-sorted mCherry+-niche
cells also showed an overall reduction in expression of alveolar lineage

44% 20%

FSC FSC

EPCAM–APC

mCherry– mCherry+

mCherry–mCherry+

Mmp9
Csf3
Wnt10a
Ccl2
Tnc
Wisp1
IL11
Wnt7b
Cxcl1
Loxl4
Loxl3
Postn

6.92
6.26
5.94
4.48
4.03
3.91
3.56
3.55
2.86
2.25
2.06
1.87

log(fold change)
mCherry+ vs
mCherry–

2,2444,8421,951

Early Late
GFP+CD45+ exclusion

Early (mCherry+ vs mCherry–)
versus
Late (mCherry+ vs mCherry–)

Single-cell suspension

Large-
metastases
(late)

Micro-
metastases
(early)

Niche RNA expression

PC1: 83% variance

PC2: 8% variance–40 –20 02040

–10
–20

0

10

mCherry– late
mCherry– early

mCherry+ late
mCherry+ early

Day 0
intraveneous
injection

Day 4 Day 5 Day 7 Day 10

Clusters

Established
micro-
metastases

Large
metastases

(early) (late)

Post-
extravasation

0.5 1.0 1.5 2.0 2.5 3.0 3.5 4.0–log (FDR q-value)

Correlation of GSEA processes with downregulated genes

Early niche

Late niche

Cilium movement
Epithelial cell differentiation
Epithelial cell development
Epithelial cell morphogenesis
Regulation of epithelial cell migration
Glandular epithelial cell differentiation
Regulation of epithelial cell migration
Cilium movement
Regulation of cell adhesion
Positive regulation of epithelial cell migration
Regulation of cell–cell adhesion
Epithelial cell differentiation in kidney development
Positive regulation of cell adhesion
Regulation of epithelial cell differentiation
Positive regulation of cell–cell adhesion
Epithelial cell differentiation
Regulation of epithelial cell proliferation

IgG controlAnti-WISP1

0

10

20

30

40

50

No. of metastases

per mouse

P = 0.05 9

In vivo metastatic
outcome

P = 0.004

0

20

40

60

80

EPCAM

+ cells (% of Lin

SP-C mCherry SP-C mCherry DAPI –)

a b

c

d e

f

g h i

Fig. 3 | The mCherry-niche strategy identifies an epithelial component
of metastatic TME. a, Schematic of metastatic progression using
labelling-4T1 cells^6. b, Experimental design for RNA-seq experiments^6.
c, Principal component analysis (PCA) of CD45−Ter119− cell signatures
from metastatic lungs at early (n = 3, 10 mice each) and late (n = 3, 5 mice
each) time points. The black oval encloses the distal lung samples; red
ovals enclose the mCherry+-niche samples to highlight their similarity
in the PCA plot. d, Venn diagram of differentially expressed genes in
the mCherry+ niche and selected factors that are common at early and
late stages. Wisp1 is also known as Ccn4. e, WISP1-blocking antibody
treatment in vivo (n = 10, 2 independent experiments). Box edges
represent 25th and 75th percentiles, the horizontal bar is the median and
the whiskers show the range of values. f, GSEA correlation from RNA-seq


data comparing early (n = 3) or late (n = 3) mCherry+ samples with their
respective mCherry− controls. g, Left, representative immunofluorescence
images of lung tissue (n = 3 mice) showing mCherry-labelled micro-
metastasis (red), SP-C (white) and DAPI (blue, middle). Right, enlarged
view of areas indicated with dashed outlines. Scale bars: main panel,
100  μm; enlarged insets, 10  μm (white arrows and dashed outlines,
mCherry-labelled SP-C+ cells). h, EPCAM+ cell frequency among Lin−
(CD45−CD31−Ter119−) cells in distal lung (mCherry−) and mCherry+
cells estimated by FACS (n = 13 mice). i, Representative FACS plots from h.
Numbers indicate the percentage of cells in the respective quadrant.
Statistical analysis by unpaired two-tailed t-test with Welch’s correction (e),
weighted Kolmogorov–Smirnov-like statistic with Benjamini–Hochberg
correction (f) and paired two-tailed t-test (h).

29 AUGUSt 2019 | VOl 572 | NAtUre | 605
Free download pdf